BRD1 Human

Bromodomain Containing 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to BRD1 Human

BRD1 (Bromodomain Containing 1), also known as BRPF2, is a nuclear protein critical for epigenetic regulation. It belongs to the BET (Bromodomain and Extra-Terminal Domain) family and functions as a chromatin reader, recognizing acetylated lysine residues on histones . This protein plays pivotal roles in transcriptional activation, chromatin remodeling, and gene expression linked to brain development and disease .

Role in Epigenetic Regulation and Disease

BRD1 regulates gene expression by modulating histone acetylation and interacting with transcription factors. Its dysregulation is implicated in psychiatric disorders and cancer.

Psychiatric Disorders

BRD1 binds near transcription start sites (TSS) of genes linked to schizophrenia and bipolar disorder . Key findings:

  • Isoform-Specific Interactions: BRD1-S interacts with 1,540 genes, while BRD1-L binds 823 genes .

  • Mitochondrial Metabolism: Modulates nuclear-encoded mitochondrial proteins (e.g., PPARα co-repressor activity), linking it to neuronal dysfunction .

  • Schizophrenia Risk: BRD1-S interacts with SUV420H1 (a histone methyltransferase) and schizophrenia-associated genes .

Cancer Implications

BRD1 is upregulated in hepatocellular carcinoma (HCC) and regulates lipid metabolism via SREBF1 (sterol regulatory element-binding protein 1) .

  • Oncogenic Role: BRD1 knockdown reduces HCC cell proliferation, migration, and tumor growth .

  • Epigenetic Mechanism: BRD1 promotes H3K9ac at SREBF1 promoters, enhancing lipogenesis and tumor progression .

Research Applications and Tools

Recombinant BRD1 proteins are essential for studying bromodomain interactions and inhibitor development.

ProductSourceTagsApplications
BRD1 (556–688)E. coli His/FLAGBinding assays, inhibitor screening
BRD1-Lenti ORF CloneHuman cell linesMyc-DDKStable expression in mammalian cells
BRD1 (561–668)E. coli HisStructural studies, SDS-PAGE analysis

Mental Health Disorders

BRD1’s interaction network is enriched with schizophrenia risk genes, particularly during fetal brain development and childhood . Isoform-specific modulation may offer therapeutic avenues.

Product Specs

Introduction
BRD1 (Bromodomain Containing 1) is a protein that contains a bromodomain, a motif often found in transcriptional coactivators. It is primarily located in the nucleus of various cell types, including testis cells. Genetic variations in BRD1 have been associated with schizophrenia and bipolar disorder.
Description
This product consists of a recombinant human BRD1 protein produced in E. coli. It is a single, non-glycosylated polypeptide chain with a molecular weight of 17.8 kDa, comprising 156 amino acids (residues 556-688). The protein has a 23 amino acid His-tag attached to its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The BRD1 protein is supplied in a solution with a concentration of 0.5 mg/ml. The solution contains phosphate buffered saline (pH 7.4), 10% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it should be frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the BRD1 protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Bromodomain Containing 1, BRL, Bromodomain And PHD Finger-Containing Protein2, BR140-Like Protein, BRPF2, Bromodomain-Containing Protein 1, Bromodomain-Containing 1, BR140-Like, BRPF1, BRD1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEQVAMEL RLTPLTVLLR SVLDQLQDKD PARIFAQPVS LKEVPDYLDH IKHPMDFATM RKRLEAQGYK NLHEFEEDFD LIIDNCMKYN ARDTVFYRAA VRLRDQGGVV LRQARREVDS IGLEEASGMH LPERPA.

Q&A

What is BRD1 and what is its primary function in human cells?

BRD1 (Bromodomain containing 1) is an epigenetic regulator that functions as a chromatin co-modifier, interacting with both proteins and DNA to control the expression of genetic networks. It plays a crucial role in transcriptional regulation and normal brain development .

At the molecular level, BRD1 targets nuclear genes encoding mitochondrial proteins and acts as a co-repressor of PPAR-mediated transcription, which is a key gate-keeper of mitochondrial metabolism . The protein primarily binds in close proximity to transcription start sites and regulates expression of numerous genes, many of which are involved with brain development and susceptibility to mental disorders .

What are the main isoforms of BRD1 and how do they differ functionally?

There are two main isoforms of BRD1 that have been extensively studied: BRD1-S (short) and BRD1-L (long) . These isoforms exhibit distinct functional properties:

  • Chromatin binding patterns: BRD1-S and BRD1-L bind to the promoter regions of 1540 and 823 genes, respectively, indicating differential targeting capabilities .

  • Protein interactions: The isoforms demonstrate some unique protein interaction partners. For example, BRD1-S specifically interacts with the histone methyltransferase SUV420H1, which has been implicated in autism through de novo mutations .

  • Transcriptional effects: Both isoforms affect gene expression when bound to promoter regions, but their specific regulatory effects may differ based on their binding partners and cellular context .

  • Mitochondrial impact: Modulation of BRD1 expression alters mitochondrial physiology, metabolism, and bioenergetics in an isoform-dependent manner .

How is BRD1 implicated in neuropsychiatric disorders?

BRD1 has been linked to several neuropsychiatric disorders through multiple lines of evidence:

  • Genetic association studies: BRD1 has been implicated in susceptibility to schizophrenia and bipolar disorder through genome-wide association studies (GWAS) .

  • Interaction networks: The BRD1 interaction network is enriched for schizophrenia risk genes, indicating its central role in pathways relevant to mental disorders .

  • Brain development: BRD1 plays a role in many brain regions throughout life, implicating regions such as the striatum, hippocampus, and amygdala at mid-fetal stages, which are critical periods for neurodevelopment related to psychiatric disorders .

  • Mitochondrial regulation: BRD1's role in modulating mitochondrial functions may be a potential mechanism linking genetic variation in BRD1 to psychopathology in humans, as mitochondrial dysfunction has been implicated in various psychiatric disorders .

What are the recommended methods for studying BRD1 protein interactions?

For investigating BRD1 protein interactions, researchers have successfully employed several complementary approaches:

  • Stable cell line generation: Creating stable human cell lines expressing epitope-tagged BRD1-S and BRD1-L provides reliable discovery systems for identifying protein interactions .

  • Co-immunoprecipitation followed by mass spectrometry: This technique has been effectively used to identify protein binding partners of BRD1 isoforms. It enabled the discovery of interactions with chromatin remodeling proteins like PBRM1 and histone modifiers such as MYST2 and SUV420H1 .

  • Protein-protein interaction networks (PPINs): Construction of PPINs based on physical interaction data from databases like BioGRID and HIPPIE can help evaluate the network characteristics of BRD1 proteins and predict their biological roles .

  • Hallmark pathway enrichment analysis: This approach, combined with clustering using gene ontology, can predict the functional characteristics of subnetworks formed by BRD1 and its interaction partners .

What experimental tools are available for manipulating BRD1 expression in research models?

Researchers can utilize various molecular tools to manipulate BRD1 expression:

  • Expression vectors: Mammalian gene expression vectors for human BRD1 are available in multiple formats, including regular plasmid, lentivirus, adenovirus, AAV, and PiggyBac vectors .

  • Knockdown vectors: shRNA lentivirus, adenovirus, AAV, and PiggyBac vectors targeting human BRD1 can be used for knockdown experiments .

  • Gene editing tools: CRISPR-based vectors (regular plasmid, lentivirus, adenovirus) are available for precise genetic manipulation of BRD1 .

  • Isoform-specific approaches: When designing experiments, it's crucial to consider the distinct functions of BRD1-S and BRD1-L isoforms, potentially requiring isoform-specific targeting strategies .

How can researchers effectively study BRD1 chromatin interactions?

To investigate BRD1 chromatin interactions, the following methodological approaches have proven valuable:

  • Chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq): This technique has been successfully used to identify genomic binding sites of both BRD1-S and BRD1-L isoforms. Results show that BRD1 primarily binds near transcription start sites of genes .

  • Integration with gene expression data: Correlating ChIP-seq data with gene expression profiles after upregulating or downregulating BRD1 can help identify genes directly regulated by BRD1 binding .

  • Isoform-specific analysis: Separate ChIP-seq experiments for BRD1-S and BRD1-L can reveal isoform-specific chromatin interactions and regulatory functions .

  • Spatiotemporal analysis: Integrating ChIP-seq data with spatiotemporal transcriptomic datasets from human brain can provide insights into the developmental and regional specificity of BRD1 function .

How does BRD1 regulate mitochondrial function, and what are the implications for neuropsychiatric disorders?

BRD1 regulates mitochondrial function through several mechanisms that may have significant implications for neuropsychiatric disorders:

  • Transcriptional regulation of nuclear-encoded mitochondrial genes: BRD1 targets nuclear genes encoding mitochondrial proteins, and modulation of BRD1 expression leads to distinct shifts in the expression profile of these genes .

  • PPAR co-repression: BRD1 acts as a co-repressor of Peroxisome proliferator-activated receptor (PPAR)-mediated transcription. PPAR is a key gate-keeper of mitochondrial metabolism, suggesting that BRD1 influences mitochondrial function through this regulatory pathway .

  • Impact on mitochondrial physiology and bioenergetics: Experimental evidence demonstrates that modulation of BRD1 expression alters:

    • Mitochondrial physiology (mtDNA content and mitochondrial mass)

    • Metabolism (reducing power)

    • Bioenergetics (basal, maximal, and spare respiration)

  • Isoform-dependent effects: These mitochondrial effects are dependent on both the expression level and the specific isoform of BRD1 (BRD1-S or BRD1-L) .

The connection between BRD1-mediated mitochondrial dysfunction and psychiatric disorders suggests a novel mechanism linking genetic variation in BRD1 to psychopathology. This could explain the cellular and atrophic changes of neurons previously associated with BRD1 deficiency and provides a potential therapeutic target for investigation .

What are the differential roles of BRD1 isoforms in chromatin regulation and how can they be studied independently?

The differential roles of BRD1 isoforms in chromatin regulation represent a complex area of investigation:

  • Distinct binding profiles: ChIP-seq studies have revealed that BRD1-S and BRD1-L bind to promoter regions of 1540 and 823 genes, respectively, indicating considerable differences in their genomic targeting .

  • Isoform-specific protein interactions: BRD1-S specifically interacts with the histone methyltransferase SUV420H1, while both isoforms share interactions with chromatin remodeling proteins like PBRM1 and histone modifiers such as MYST2 .

  • Methodological approaches for independent study:

    • Generate isoform-specific expression constructs with unique epitope tags

    • Develop isoform-specific antibodies when possible

    • Design siRNA or CRISPR strategies targeting unique regions of each isoform

    • Use isoform-specific ChIP-seq followed by differential binding analysis

    • Employ isoform-specific rescue experiments in knockdown models

  • Functional validation: After identifying isoform-specific binding sites, researchers should validate the functional consequences through targeted gene expression analysis, histone modification profiling at specific loci, and assessment of downstream cellular processes .

How can the BRD1 interaction network be integrated with clinical data to advance understanding of psychiatric disorders?

Integrating the BRD1 interaction network with clinical data offers promising avenues for psychiatric disorder research:

  • GWAS data integration: The BRD1 interaction network has been shown to be enriched for schizophrenia risk genes. Researchers can further leverage this by systematically integrating protein interaction data with results from large-scale GWAS studies of various psychiatric disorders .

  • Exome sequencing data analysis: Rare variants identified in psychiatric disorder cohorts can be mapped onto the BRD1 interaction network to identify potentially disrupted pathways. This approach has already yielded insights, such as the identification of de novo mutations in SUV420H1 (a BRD1-S interaction partner) in autism .

  • Spatiotemporal transcriptomic integration: The BRD1 interaction network is predominantly co-expressed with BRD1 mRNA in specific regions of the human brain during development. Integrating this information with clinical neuroimaging or postmortem studies can help identify critical developmental windows and brain regions for therapeutic intervention .

  • Pathway-based approaches: The BRD1 interaction network is enriched for pathways involved in gene expression and brain function. Researchers can leverage this by conducting pathway-based analyses of clinical data to identify patient subgroups with potential BRD1-related dysregulation .

What are the key considerations when designing experiments to investigate BRD1 function in different cell types?

When investigating BRD1 function across different cell types, researchers should consider:

  • Endogenous expression profiles: Characterize the endogenous expression levels of BRD1 isoforms in the cell type of interest, as this may influence the interpretation of overexpression or knockdown experiments .

  • Isoform ratios: The ratio between BRD1-S and BRD1-L may vary by cell type and impact cellular function. Quantitative PCR or western blotting with isoform-specific antibodies should be used to assess these ratios .

  • Interaction partner expression: Key BRD1 interaction partners like PBRM1, MYST2, and SUV420H1 may be differentially expressed across cell types. Their presence or absence could significantly alter BRD1 function .

  • Developmental timing: For neural cell types in particular, the developmental stage may influence BRD1 function, as BRD1's role in brain development appears stage-specific across different regions .

  • Functional readouts: Select appropriate functional assays based on the cell type. For neural cells, consider mitochondrial function, gene expression profiling, and morphological analyses. For non-neural cells, focus on general chromatin regulation and cell-type-specific functional outcomes .

How can researchers address the challenges of studying BRD1 in the context of neurodevelopment and psychiatric disorders?

Studying BRD1 in neurodevelopment and psychiatric disorders presents unique challenges that can be addressed through:

  • Model system selection: Consider using:

    • Patient-derived induced pluripotent stem cells (iPSCs) differentiated into relevant neural lineages

    • Transgenic mouse models with conditional BRD1 knockout or isoform-specific expression

    • Brain organoids to recapitulate neurodevelopmental processes in a three-dimensional context

  • Temporal dynamics: Implement time-course experiments that capture different neurodevelopmental stages, as BRD1's impact appears to be stage-specific .

  • Region-specific analyses: Focus on brain regions implicated in psychiatric disorders and showing high BRD1 expression, such as striatum, hippocampus, and amygdala at mid-fetal stages .

  • Multi-omics approaches: Integrate transcriptomics, proteomics, and epigenomics data to comprehensively characterize BRD1's impact on neural development and function .

  • Functional networks: Analyze the BRD1 interaction network in the context of psychiatric disorder risk genes to identify convergent pathways that might represent therapeutic targets .

What are the recommended approaches for interpreting contradictory findings in BRD1 research?

When faced with contradictory findings in BRD1 research, consider the following systematic approaches:

  • Isoform-specific effects: Determine whether contradictions might be explained by different BRD1 isoforms being studied, as BRD1-S and BRD1-L have distinct functions and interaction partners .

  • Cell type and context dependence: Evaluate whether different cell types or experimental conditions might explain contradictory results, as BRD1 function may vary considerably across cellular contexts .

  • Expression level considerations: Assess whether overexpression versus endogenous expression versus knockdown conditions might contribute to seemingly contradictory outcomes .

  • Methodology differences: Carefully compare experimental methods, including:

    • Antibody specificity and epitope accessibility

    • ChIP-seq protocols and peak calling algorithms

    • Cell synchronization status and cell cycle effects

    • Data normalization approaches

  • Integrated analysis: When possible, perform integrated analyses of contradictory datasets using standardized bioinformatic pipelines to identify potential sources of variation and core consistent findings .

What emerging technologies could advance our understanding of BRD1 function in human brain development?

Several cutting-edge technologies hold promise for advancing BRD1 research:

  • Single-cell multi-omics: Combining single-cell transcriptomics, proteomics, and epigenomics can reveal cell-type-specific functions of BRD1 during brain development and in psychiatric disorders.

  • CRISPR-based epigenome editing: Targeted modification of epigenetic marks at BRD1 binding sites can help elucidate the causal relationship between BRD1 binding and gene expression changes.

  • Spatial transcriptomics: These techniques can map BRD1-regulated gene expression in intact brain tissue, providing spatial context to transcriptional changes.

  • Live-cell imaging of chromatin dynamics: Visualizing BRD1 isoform recruitment to chromatin in real-time can reveal dynamic aspects of its function that are missed in static analyses.

  • Brain organoid models with genetic modifications: Creating brain organoids with BRD1 mutations or isoform-specific alterations can model neurodevelopmental aspects of psychiatric disorders in a human cellular context.

How can systems biology approaches enhance our understanding of BRD1's role in psychiatric disorders?

Systems biology approaches offer powerful frameworks for understanding BRD1's complex role:

  • Network medicine: Analyzing BRD1 within protein-protein interaction networks can identify its position in disease modules related to psychiatric disorders .

  • Pathway integration: Integrating BRD1-regulated genes with known psychiatric disorder pathways can reveal convergent mechanisms and potential points for therapeutic intervention .

  • Multi-scale modeling: Developing computational models that span from molecular interactions to cellular and circuit-level phenotypes can help predict the emergent effects of BRD1 dysfunction.

  • Pharmacological network perturbation: Systematic analysis of how psychiatric medications affect the BRD1 interaction network could reveal mechanisms of action and suggest novel therapeutic approaches.

  • Cross-disorder analysis: Examining BRD1's role across multiple psychiatric disorders may reveal shared pathophysiological mechanisms and explain clinical comorbidities.

Product Science Overview

Introduction

Bromodomain Containing 1 (BRD1) is a protein encoded by the BRD1 gene in humans. This protein is part of the bromodomain family, which is known for its role in recognizing acetylated lysine residues on histone tails, thereby playing a crucial role in the regulation of gene expression.

Structure and Function

BRD1 contains a bromodomain, an approximately 110 amino acid protein domain that recognizes acetylated lysine residues. This recognition is essential for protein-histone association and chromatin remodeling . The bromodomain adopts an all-α protein fold, forming a hydrophobic pocket that specifically binds to acetylated lysine .

BRD1 is a component of the MOZ/MORF acetyltransferase complex, which stimulates the acetylation of histones H3 and H4. This acetylation is a key step in gene activation, as it loosens the chromatin structure, allowing transcription factors to access DNA . BRD1 plays a significant role in hematopoiesis by directing the specificity of the HBO1 complex towards histone H3 ‘Lys-14’ acetylation, promoting erythroid differentiation .

Clinical Significance

Variations in the BRD1 gene have been associated with several psychiatric disorders, including schizophrenia and bipolar disorder . The protein’s role in gene activation and chromatin remodeling suggests that it may influence the expression of genes involved in these conditions.

Recombinant BRD1

Human recombinant BRD1 is produced in E. coli and is a single, non-glycosylated polypeptide chain containing 156 amino acids. It has a molecular mass of approximately 17.8 kDa and is fused to a 23 amino acid His-tag at the N-terminus for purification purposes .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.